WO2014120922A1 - Gel de silice poreux comme support pour des technologies liquides - Google Patents

Gel de silice poreux comme support pour des technologies liquides Download PDF

Info

Publication number
WO2014120922A1
WO2014120922A1 PCT/US2014/013848 US2014013848W WO2014120922A1 WO 2014120922 A1 WO2014120922 A1 WO 2014120922A1 US 2014013848 W US2014013848 W US 2014013848W WO 2014120922 A1 WO2014120922 A1 WO 2014120922A1
Authority
WO
WIPO (PCT)
Prior art keywords
inorganic oxide
composition according
oxide particles
active ingredient
biologically active
Prior art date
Application number
PCT/US2014/013848
Other languages
English (en)
Inventor
Frederik Hendrik MONSUUR
Original Assignee
W. R. Grace & Co.-Conn.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by W. R. Grace & Co.-Conn. filed Critical W. R. Grace & Co.-Conn.
Priority to ES14746507T priority Critical patent/ES2908958T3/es
Priority to CN201480019478.7A priority patent/CN105102117A/zh
Priority to PL14746507T priority patent/PL2950923T3/pl
Priority to KR1020157023665A priority patent/KR102229036B1/ko
Priority to US14/764,363 priority patent/US10660856B2/en
Priority to EP14746507.4A priority patent/EP2950923B1/fr
Priority to JP2015556128A priority patent/JP6513031B2/ja
Publication of WO2014120922A1 publication Critical patent/WO2014120922A1/fr
Priority to HK16105942.0A priority patent/HK1217927A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/143Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with inorganic compounds
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/02Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material
    • B01J20/10Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material comprising silica or silicate
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/02Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material
    • B01J20/10Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material comprising silica or silicate
    • B01J20/103Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof comprising inorganic material comprising silica or silicate comprising silica
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/28Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties
    • B01J20/28002Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties characterised by their physical properties
    • B01J20/28004Sorbent size or size distribution, e.g. particle size
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/28Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties
    • B01J20/28054Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties characterised by their surface properties or porosity
    • B01J20/28057Surface area, e.g. B.E.T specific surface area
    • B01J20/28059Surface area, e.g. B.E.T specific surface area being less than 100 m2/g
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/28Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties
    • B01J20/28054Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof characterised by their form or physical properties characterised by their surface properties or porosity
    • B01J20/28069Pore volume, e.g. total pore volume, mesopore volume, micropore volume
    • B01J20/28073Pore volume, e.g. total pore volume, mesopore volume, micropore volume being in the range 0.5-1.0 ml/g
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • CCHEMISTRY; METALLURGY
    • C01INORGANIC CHEMISTRY
    • C01BNON-METALLIC ELEMENTS; COMPOUNDS THEREOF; METALLOIDS OR COMPOUNDS THEREOF NOT COVERED BY SUBCLASS C01C
    • C01B33/00Silicon; Compounds thereof
    • C01B33/113Silicon oxides; Hydrates thereof
    • C01B33/12Silica; Hydrates thereof, e.g. lepidoic silicic acid
    • C01B33/18Preparation of finely divided silica neither in sol nor in gel form; After-treatment thereof
    • C01B33/187Preparation of finely divided silica neither in sol nor in gel form; After-treatment thereof by acidic treatment of silicates
    • C01B33/193Preparation of finely divided silica neither in sol nor in gel form; After-treatment thereof by acidic treatment of silicates of aqueous solutions of silicates

Definitions

  • the present invention relates to compositions comprising inorganic oxide porous material containing a biologically active ingredient in liquid form, methods of making such compositions; and methods of using them.
  • the oral route remains the preferred route of drug administration due to its convenience and good patient compliance.
  • Major problems in oral drug formulations are the erratic and incomplete absorption throughout the gastrointestinal (GI) tract, resulting in low and variable bioavailabilit ' and Sack of dose proportionality. These problems mainly result from poor aqueous solubility of the active ingredient, it has been reported that an estimated 40% of existing pharmaceutical active ingredients and an even higher proportion of all newly developed drugs are poorly soiubie or insoluble in water. This poses a major challenge to drug development, as there is a high need for producing suitable formulations to improve the solubility and bioavailability of such drugs.
  • Methods that have been developed include the reduction of particle size of the drug by micronisation or nanonisaiion as to increase surface area, thereby increasing dissolution rate of the active ingredient. Further methods include solubilization in surfactant systems, water- soluble molecular complexes with cyelodextrins, converting the drug in amorphous form by lyophilization or formation of solid dispersions in hydrophiiie carriers, microencapsulation, and the release from porous carrier materials.
  • Hpid based drug delivery system LBDDS
  • the active ingredient is already in solution so that the drug is present on molecular level, avoiding the dissolution step from the crystalline state.
  • the drugs in liquid phase are typically fdled into soft gelatin capsulees.
  • iiquisoiid formulations which are porous carrier materials wherein the drugs remain in liquid form.
  • Liquisolid forms are obtained by conversion of drugs in liquid form into acceptably flowing non-adherent and compressible powder mixtures by blending with selected carriers and coating materials. These then are converted into solid dosage forms such as tablets, pellets, and capsulees.
  • Iiquisoiid dosage forms of water insoluble drugs show improved dissolution properties and bioavailability. This technique was successfully applied for low dose water-insoluble drugs.
  • loadability and release of the drugs front the carriers used is limited, formulation of insoluble drags at higber doses is one of the limitations of the Iiquisoiid technique.
  • Another problem associated with iiquisoiid formulations is their decreased flowability when loaded with higher amounts of drugs in liquid form. This causes these materials difficult to process in pharmaceutical manufacturing. In order to have acceptable flowability and compactabihty, high levels of carrier and coating materials have to be added thereby increasing the weight and volume of the resulting dosage forms,
  • SEDDS self-emulsifying drug delivery systems
  • This type of emulsion-based drug formulations can be used in soft gelatin capsulees or as Iiquisoiid formulations.
  • SEDDS are isotropic and thermodynamically stable mixtures of drug, oil lipid, surfactant/cosurfactant, thai, in contact with aqueous fluids, spontaneously form oil-in-water emulsions of droplets, ranging in droplet size approximately between 100-300 nrn.
  • SMEDDS self-micro-emulsifying drug delivery systems
  • SNEDDS seif-nanoemulsifying drug delivery system
  • SEDDS, SNEDDS and SMEDDS have also been converted into solid-SEDDS, solid-SNEDDS or soHd-SMEDDS (S-SEDDS, S-5MEDDS or S-SMEDDS) using liquisoiid solidification procedures similar as described above.
  • S-SEDDS, S-5MEDDS or S-SMEDDS solid-SEDDS
  • S-SMEDDS solid-SNEDDS
  • soHd-SMEDDS S-SEDDS, S-5MEDDS or S-SMEDDS
  • the resulting solid formulations in turn can be worked into various solid or semisolid dosage forms (tablets, pellets, capsules, creams, transdermal systems, etc).
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having a pore volume, as measured by nitrogen porosirnetry, of about 0.5 cm 3 /g or greater; wherein the inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1.5: 1, or 1.6: 1 , or 1.7: 1 , or 1.8: 1, or 1.9: 1, up to 2: 1.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cnr ' g or greater; wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1.5 : i , or 1.6: 1 , or 1.7: 1 , or 1.8: 1 , or 1.9: 1 , up to 2: 1.
  • the present invention relates to a method of making composition
  • a method of making composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/ 100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 em 3 /g or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1 ,5: 1 , or 3 ,6: 1, or 1.7: 1, or 1 .8: 1. or 1 .9: 1 , up to 2: 1.
  • the present invention concerns a method of making a pharmaceutical and/or cosmetic composition
  • a pharmaceutical and/or cosmetic composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a Hquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cnv'/g or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1.5: 1, or 1 ,6: 1 , or 1 .7: 1 , or 1 ,8: 1 , or 1 ,9: 1 , or at least 2: 1 .
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 m 1/100 g; (b) pores having a pore volume, as measured by nitrogen porcsmietry, of about 0.5 cm 3 /g or greater; and (c) a pore size distribution having a relative span of about 1.5 or less, or about 1.4 or less, or about 1 ,3 or less, or about 1.2 or less, or about 1.1 or less, or about 1 .0 or less,
  • the present invention concerns a composition
  • a composition comprising porous Inorganic oxide particles containing a biologically active ingredient in a Hquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cm ' Vg or greater; and (c) a median pore size of 5 nm to 30 tint, wherein said inorganic oxide particles remain free flowing at a ratio of Hquid material to inorganic oxide particles of at least 1 : 1, or 1.3, or 1 ,5: 1, or 1 ,6: 1 , or 1.7: 1, or 1.8: 1 , or 1.9: 1, or at least 2: 1.
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm " g or greater; and (c) a median particle size of from 3 microns to 10 mm; wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1 : 1 , or 1 ,3, or 1.5: 1 , or 1 ,6: 1 , or 1.7: 1 , or 1.8: 1. or 1.9: 1 , or at least 2: 1.
  • the present invention concerns a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0.5 cnrVg or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material, decreases in volume of at least about 15% after resting, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40% after resting.
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cnrVg or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material, increases in bulk density by at least about 35% after resting, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%, or at least about 40% after resting.
  • the present invention concerns a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cm ' Vg or greater; wherein, after mixing the inorganic oxide particles and liquid material and then resting for at least 2 hours, at least about 400 mg of said composition may be loaded into a zero size capsule.
  • at least about 410 mg, or at least about 420 trig, or at least about 430 rug of said composition may be loaded into a zero s e capsule.
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a Hquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 enr/g or greater; and (c) a ratio of liquid material to inorganic oxide particles of at least 1 : 1 ; wherein at least 65% of the liquid material is desorbed from the particles upon desorption.
  • the inorganic oxide material comprises porous particles comprising pores having a mean pore diameter in the range of about 5 nm to about 30 nm.
  • the porous inorganic oxide material containing a biologically active ingredient in a liquid material is in the form of particles, i.e. particles of porous inorganic oxide material containing a biologically active ingredient in liquid form.
  • the average diameter of the particles of porous inorganic oxide material of the invention may be in the range of from about 3 ⁇ to about 5 mm.
  • compositions of the invention may contain further liquid organic auxiliary materials such as oils, non-volatile solvents, and surfactants.
  • the particles of porous inorganic oxide material containing a biologically active ingredient in a Hquid material of the invention form a powder that is free-flowing.
  • the powder has a Carr index of equal or lower than 25, or the powder has a Hausner index of about 1 to about 1 ,4.
  • the angle of repose of said powder is from about 30 i! to about 45 3 ⁇ 4 , These properties are measured before and after loading the biologically active ingredient on the porous inorganic oxide material.
  • the composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material has a bulk bulk density, after loading and resting during at least 2 hours, of at least 450 g/1.
  • the resulting loaded porous Inorganic oxide material may show a limited (e.g. up to about 10%) or no increase, but in particular shows a decrease in volume when compared to the unloaded inorganic oxide porous material In one embodiment the decrease is up to about 30%, or up to about 20%, or up to about 10% (each % in this paragraph being volume/volume or v/v).
  • a limited (e.g. up to about 10%) or no increase but in particular shows a decrease in volume when compared to the unloaded inorganic oxide porous material
  • the decrease is up to about 30%, or up to about 20%, or up to about 10% (each % in this paragraph being volume/volume or v/v).
  • Each of the changes in volume and/or density mentioned herein are after loadsng and resting up to 2 hours depending upon the liquid, and even up to 24 or 48 hours.
  • a decrease of the volume or increase in density in particular the particular decreases of volume or increases in density mentioned herein, during a time period of about 3 hrs, in particular of about 2 hrs Is observed, after which time period no further decrease occurs and the volume stays substantially constant.
  • compositions of the present invention advantageously do not .show the substantial increase of volume that is typically observed when loading art-known silicas with a biologically active ingredient in a liquid material This allows the manufacture of more compact dosage forms, which in turn contributes to better processabllity during pharmaceutical manufacturing and to better acceptability of the dosage forms by patients,
  • the present invention concerns a pharmaceutical composition
  • a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material, wherein the inorganic o ide material has the oil adsorption, the pore volume, and BET surface area, as specified herein, in one embodiment the said composition comprising inorganic oxide porous material containing a biologically active ingredient is in the form of particles.
  • the present invention is further directed to methods of making the disclosed compositions, in one embodiment, the method of making a composition of the present invention comprises incorporating at least one biologically active ingredient in a liquid material into the porous inorganic oxide material having the oil adsorption, the pore volume, and BE T surface area, as specified herein.
  • the present invention concerns a method of making a pharmaceutical composition
  • a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material, wherein the inorganic oxide material has the oil adsorption, the pore volume, and BET surface area, as specified herein, said method comprising combining said pharmaceutical dosage formulating ingredient with said composition.
  • the present invention is also directed to methods of using the disclosed compositions, in one embodiment, the method of using a composition of the present invention comprises administering a composition of the invention to a patient so as to deliver a biologically active material to the patient, wherein the composition comprises a porous inorganic oxide material containing a biologically active ingredient n a liquid material, wherein the inorganic oxide material has the oil adsorption, the pore volume, and BET surface area, as specified herein.
  • the composition that is administered in particular is a pharmaceutical dosage form.
  • the said inorganic oxide material is in the form of particles, in particular particles havin an average diameter as further specified herein.
  • FIG. 1 graphically displays the kinetic behaviour or volume change over time of an esemplasy composition of the present invention
  • FIG. 2 graphically displays capsule loading capacity change before and after resting of an exemplary composition of the present Invention according to Example 4.
  • FIG. 3 graphically displays a release profile of acetaminophen from an exemplary composition of the present invention according to Example 6.
  • FIG. 4 graphically displays a release profile of ascorbic acid from an exemplary composition of the present, invention according to Example 6.
  • FIG, 5 graphically displays the release profile Gyburide from a solid SEDDS system of the present invention according to Example 8.
  • biologically active ingredient means an active pharmaceutical ingredient (API), which provides a pharmacological activity or otherwise have direct effect in the diagnosis, cure, mitigation, treatment or prevention of disease, or to have direct effect In restoring, correcting or modifying physiological functions in humans. Even though this includes poorly soluble material, it may also include materials that range in solubility, including those listed in the BCS (Biopharmaceutic Classification System), which is a classification approach where drags (APIs) are divided into four classes based on the extent (high or low) of their aqueous solubility and permeability through the GI tract wail in particular intestinal.
  • API active pharmaceutical ingredient
  • these four classes are: (Group I) High Solubility and High Permeability drags, (Group ⁇ ) Low Solubility and High Permeability drugs, (Group ⁇ ) High Solubility and Low Permeability drugs and, (Group IV) Low solubility and Low Permeability drugs.
  • bulk density means the mass of particulate matter, such as a powder, divided by the total volume occupied by the matter, and includes mtraparticle pore volume and interpariicle void volume,
  • the term "free flowing” means the ability of a group of particles or a powder to move when a force (e.g., gravity, or other external force) is applied to it.
  • a force e.g., gravity, or other external force
  • Commonly used tests for measuring powder flow include the Hausner ratio, compressibility index (Carr index) or angle of repose.
  • the compressibility index (Can index) and the Hausner ratio are determined by measurin both the bulk volume and the tapped volume of a powder.
  • inorganic oxides is defined as binary oxygen compounds where the inorganic component is the cation and the oxide is the anion.
  • the inorganic material includes metals may also include metalloids. Metals include those elements on the left of the diagonal line drawn from boron to polonium on the periodic table. Metalloids or semi-metals include those elements that are on the right of this line. Examples of inorganic oxides include silica, alumina, titania, zirconia, etc., and mixtures thereof.
  • the term "intraparticle pore volume” means pore volume attributable to the spaces in the pore of the particles, as compared to interparticle pore volume, which is the pore volume attributable to the spaces between the particles (i.e., the interstitial space).
  • lipid materia! or “lipid component” means organic materials comprising fatty acids and their derivatives, and substances related biosyntheticaiiy or functionally to these compounds. They include, but are not limited to, molecules that originate entirely or in part by carban ion-based condensations of thioesters (fatty acids, po!yketides, etc) and/or by carocation-based condensations of isoprene units (preno!s, sterols, etc.).
  • ordered porous material' ' ' refers to porous particles that have an interna! structural order such that they possess a low angle X-ray diffraction patterns according to Bragg's Law.
  • ordered mesoporous silica for example, MCM-41 , SBA- 15, TUD-1 , HMM-33 and FSM- 56,
  • non-ordered porous material refers to porous particles possessing an internal structure such that they do not have a low angle X-ray diffraction pattern according to Bragg' s Law, Such materials may be formed via any known process including, but not limited to, a solution polymerization process such as for forming colloidal particles, a continuous flame hydrolysis technique such as for forming fused particles, a gel technique such as for forming gelled particles, and a precipitation technique such as for forming precipitated particles. The particles may be subsequently modified by autoclaving, flash drying, super critical fluid extracting, etching, or like processes.
  • the particles may be composed of organic and/or inorganic materials and combinations thereof, in one exemplar ⁇ ' embodiment the particles are composed of inorganic materials such as inorganic oxides, sulfides, hydroxides, carbonates, silicates, phosphates, etc, hut are preferably inorganic oxides.
  • the particles may be a variety of different symmetrical, asymmetrical or irregular shapes, including chain, rod or lath shape.
  • the particles may have different structures including amorphous or crystalline, etc.
  • the particles may include mixtures of particles comprising different compositions, sizes, shapes or physical structures, or that may be the same except for different surface treatments, Porosiiy of the particles may be intraparticle or interparticle in cases where smaller particles are agglomerated to form larger particles.
  • the particles are composed of inorganic materials such as inorganic oxides, sulfides, hydroxides, carbonates, silicates, phosphates, etc, hut are preferably inorganic oxides.
  • Porous materials include organic and inorganic materials, or hybrids thereof, and may be in the form of particles, monoliths, membranes, coatings, and the like.
  • pore size distribution means the relative abundance of each pore size in a representative volume of porous inorganic particles.
  • media pore size is the pore diameter below which 50% of the intraparticle pore volume resides.
  • the term "relative span” is defined as meaning a measure of the breadth of pore size distribution.
  • the “span” is measured by subtracting the d3 ⁇ 4o pore size (i.e., the pore size/diameter below which 10% of the pore volume resides) from the d3 ⁇ 43 ⁇ 4 pore size (i.e., the pore size/diameter below which 90% by pore volume resides) as measured by mercury porosimetry.
  • the term “relative span” is defined as the ratio of (d3 ⁇ 4rdto)/dso. he span and relative span are determined using nitrogen sorption (BJH method) of the cumulative pore volume.
  • the term "rested ' ' or “after resting” is used to indicate a period of time wherein the porous inorganic oxide materia! is allowed to stand undisturbed after loading with a biologically active ingredient in a liquid material.
  • w/w means weight/weight and w/v means weight/volume.
  • the present invention is directed to compositions comprising a biologically active material or ingredient and an inorganic oxide material, wherein the inorganic oxide material comprises porous particles. Efficient loading of biologically active materials in liquids or a liquid material onto vehicles for drug delivery is a concern for many biologically active materials and this invention relates to various embodiments that provide solutions to this problem, Applicants of the present invention have found that certain porous inorganic oxide materials having a specific sets of physical properties provide exceptional liquid loading and release of biologically active materials that are in various liquids or a liquid material
  • the present invention reiates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particies possess (a) an oil adsorption of about 100 to about 500 mi/100 g; and (h) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm3 ⁇ 4 or greater; wherein the inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1 ,5: 1 , or 1 .6:1 , or 1 ,7: 1 , or 1.8: 1, or 1.9: 1 , up to 2: 1.
  • the present invention relates to a method of making composition
  • a method of making composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having a pore volume * as measured by nitrogen porosimetry, of about 0,5 enrVg or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1 .5: 1 , or 1.6: 1 , or 1.7: 1 , or 1 ,8: 1, or 1 ,9: 1, up to 2: 1.
  • the present " invention concerns a method of making a pharmaceutical composition
  • a pharmaceutical composition comprising at least one pharmaceutical dosage formulating ingredient and a composition comprising porous inorganic oxide particies containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi 100 g; and (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm J g or greater; wherein said inorganic oxide particles remain free flowing at a weight ratio of liquid material to inorganic oxide particles of at least 1.5: 1 , or 1.6: 1, or 1.7: 1, or 1.8: 3 , or 1.9: 1, or at least 2: 1.
  • the present invention concerns a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 300 to about 500 ml/1 GO g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm 3 /g or greater; and (e) a median pore size of 5 nm to 30 nm, wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 3 : 1 , or 13, or 1 ,5: 1, or 1.6: 1 , or 1.7: 1 , or 1 .8: 1 , or 1 ,9: 1 , or at least 2: 1.
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/ 100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm Vg or greater; and (c) a median particle size of from 3 microns to 10 mm; wherein said inorganic oxide particles remain free flowing at a ratio of liquid material to inorganic oxide particles of at least 1 : 1 , or 1 ,3. or 1 .5: 1 , or 1.6: 1 , or 1 ,7: 1, or 1.8: 1 , or 1.9: L or at least 2: 1 .
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 mi/100 g; (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 eirf/g or greater; and (c) a pore size distribution where at least 0.5 cm 3 /g of pore volume are from pores ranging from 10 nm to 30 nm, at least 0.5 enrVg, at least 0.6 cm J /g , at least 0.8 cnrVg* at least 1.0 cm Vg, at least 1 ,2 cmVg, at least 1.4 cm ' Vg, or at least 1.6 cm 7g of pore volume arc from pores ranging from 10 nm to 30 nm,
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; (b) pores having a pore volume, as measured by nitrogen porosimetry, of about 0,5 cm 3 /g or greater ; and (c) a pore size distribution having a relative span of about 1.5 or less, or about 1 ,4 or less, or about 1.3 or less, or about 1.2 or less, or about 1.1 or less, or about 1.0 or less.
  • the porous inorganic oxide material containing a biologically active ingredient in a liquid material may be in the form of particles, which may have a variety of different symmetrical, asymmetrical or irregular shapes, including chain, rod or lath shape.
  • the particles may include mixtures of particles comprising different compositions, sizes, shapes or physical structures,
  • the average diameter of the particles of the porous inorganic oxide material of the invention may be in the range of from about 3 pm to about 5 mm, preferably from about 50 ⁇ (or about 44 ⁇ ) to about 500 pm; or from about 70 pm (or about 74 pm) to about 200 ⁇ ; or from about 50 pm (or about 44 ⁇ ) to about 350 pm (or about 149 pm); or from about 50 pm (or about 44 pm) to about 150 pm (or about 149 ⁇ : or from about 100 ⁇ (or about 105 ⁇ ) to about 120 ⁇ (or about 125 ⁇ ), or from about 48 pm (or about 44 pm) to about 65 ⁇ (or about 63 pm), or from about 90 ⁇ to about 130 ⁇ .
  • the average diameters disclosed herein preferably are determined by Malvern instrumentation. The desired particle sizes can be obtained by milling and subsequent mesh sieving,
  • the porous inorganic oxide materials may be formed via any known process including a solution polymerization process such as for forming colloidal particles, a continuous flame hydrolysis technique such as for forming fused particles, a gel technique such as for forming gelled particles, and a precipitation technique such as for forming precipitated particles.
  • the particles may be subsequently modified by autociaving, flash drying, super critical fluid extracting, etching, or like processes.
  • the particles are composed of inorganic materials such as inorganic oxides, sulfides, hydroxides, carbonates, silicates, phosphates, etc, hut are preferably inorganic oxides.
  • the particles may include mixtures of particles comprising different compositions, sizes, shapes or physical structures, or that may be the same except for different surface treatments,
  • Porous materials include organic and inorganic materials, or hybrids thereof and may be in the form of particles, monoliths, membranes, coatings, and the like.
  • porous inorganic oxide material that is suitable for use in the present invention includes precipitated inorganic oxide particles and inorganic oxide gel particles. These inorganic oxides are referred to herein as “parent inorganic oxides", “parent particles”, or “parent dispersions”.
  • the porous inorganic oxide materials are non-ordered, and may further possess random intraparticle porosity. Such materials have been found, when combined with the biologically active ingredient and liquid material, to provide beneficial adsorption and desorpiion (and dissolution) characteristics. Even though any inorganic oxide composition may be suitable for use in this invention (e.g., Si0 2 , AI2O3, AIPO4, MgO, TiOz, ZrO? etc.), one embodiment of the present invention includes amorphous precipitated silica and siliea gel.
  • the inorganic oxides may also include mixed inorganic oxides such as SiC A ⁇ Os, MgO.SiO2.AbO 3 and the like.
  • Mixed inorganic oxides are prepared by conventional blending or coge!ling procedures, in embodiments comprising gels, the dispersions are derived from porous inorganic oxide geis such as gels comprising Si0 2 , A !;.(),, A I FO4, MgO, TiOj, and ZrQ 2 ,
  • the gels can be hydrogeis, aerogels, or xerogels.
  • the inorganic oxide gels include a non-ordered porous silica gel that includes interpartiele pore volume.
  • a silica gel may be prepared by mixing an aqueous solution of an alkali metal silicate (e.g., sodium silicate) with a strong acid such as nitric or sulfuric acid, the mixing being done under suitable conditions of agitation to form a clear silica sol which sets into a hydrogel, i.e., macroge!, in less than about one-half hour. The resulting gel is then washed.
  • an alkali metal silicate e.g., sodium silicate
  • a strong acid such as nitric or sulfuric acid
  • the concentration of inorganic oxide, i.e., SiC ., formed in the hydrogel is usually in the range of about 10 and about 50, or between about 20 and about 35, or between about 30 and about 35 weight percent, with the pH of that gel being from about 1 to about 9, or 1 to about 4.
  • a wide range of mixing temperatures can be employed, this range being typically from about 20 to about 50 ⁇ C.
  • the newly formed hydrogeis are washed simply by immersion in a continuously moving stream of water, which leaches out the undesirable salts, leaving about 99.5 weight percent or more pure inorganic oxide behind.
  • the pH, temperature, and duration of the washing will influence the physical properties of the siliea, such as surface area (SA) and pore volume (PV).
  • Silica gei washed at 65-90" C.
  • inorganic oxide gels such as alumina and mixed inorganic oxide gels such as siiiea/alismma cogels are also well known in the art, such as by conventional blending, co-gelation, co-precipitation, and the like. Methods for preparing such geis have been described in US 4,226,743.
  • alumina gels are prepared by mixing alkali rnetai aluminaies and aluminum sulfate
  • Cogels are prepared by cogeliing two or more metal oxides so that the geis are composited together.
  • silica alumina cogels can be prepared by gelling an alkali metal silicate with an acid or acid salt, and then adding alkali metal aluminate, aging the mixture and subsequently adding aluminum sulfate. The gel is then washed using conventional techniques.
  • Another embodiment of this invention is derived from dispersions of certain precipitated inorganic oxides. Reinforced precipitated silica such as that described in US 4,157,920 can also be used to prepare the dispersion of this invention.
  • reinforced precipitated silicas can be prepared by first acidulating an alkali inorganic silicate to create an initial precipitate. The resulting precipitate is then reinforced or "post conditioned" by additional silicate and acid. The precipitate resulting from the second addition of silicate and acid comprises 10 to 70% by weight of the precipitate initially prepared. It is believed that the reinforced structure of this precipitate is more rigid than conventional precipitates as a result of the second precipitation,
  • a liquid phase of the selected inorganic oxide is prepared.
  • the parent dispersion should be in a state that can be wet milled.
  • the medium for the liquid phase can be aqueous or non-aqueous, e.g., organic.
  • the liquid phase can he residual water in inorganic oxide gels which have been drained, but not yet dried, and to which additional water is added to reslurry the gel.
  • dried inorganic oxides e.g., xerogels
  • the parent dispersion is then milled.
  • the milling is conducted "wet", i.e., in liquid media.
  • the general milling conditions can vary depending on the feed material, residence time, impeller speeds, and milling media particle size. The techniques for selecting and modifying these conditions to obtain the desired dispersions are known to those skilled in the art.
  • the milling equipment used to mill the parent inorganic oxide particles should be of the type capable of severely milling and reducing materials to particles having the desired size, e.g., through mechanical action.
  • Such mills are commercially available, with fluid energy mills, hammer mills, and sand mills being particularly suitable for this purpose.
  • Hammer mills impart the necessary mechanical action through high speed metai blades
  • sand mills impart the action through rapidly churning media such as zirconia or sand beads.
  • Impact mills can also be used.
  • milling is not needed, such as for air-set inorganic oxide gels.
  • Such gels are formed by air- spraying an intimate mixture of an alkali metal solution (e.g., sodium silicate) with a suitable acid (e.g.. sulfuric acid) at such a concentration so that mixture gels during flight, before being collected in a suitable medium, generally water.
  • an alkali metal solution e.g., sodium silicate
  • a suitable acid e.g. sulfuric acid
  • any resulting dispersion or powder may also be further processed.
  • further processing is desirable if there is a need to prepare a relatively stable dispersion without the aid of dispersing agents, or if there is a significant population of particles that are larger than desired. Further processing may also be needed to insure that essentially all of the distribution of particles is below a certain size.
  • the dispersion or powder is processed to separate the smaller particles from the larger particles. This separation can be created by centrifuging the inorganic oxide particles into a supernatant phase, which comprises the smaller particles of the final product and a settled phase which comprises the larger particles. The supernatant phase is then removed from the settled phase, e.g., by decanting.
  • the supernatant two, three or more times to further remove large particles remaining after the initial centrifuge. It is also contemplated that the larger particles of a milled dispersion can separate over time under normal gravity conditions, and the supernatant can be removed by decanting. Depending on the product particle size targets, the settled phase also can be regarded as particles of this invention.
  • the dispersion of particles or powder also can be modified after milling to insure a stable dispersion. This can be accomplished through pH adjustment, e.g., adding alkaline material, or by the addition of conventional dispersants.
  • the inorganic oxide material in the compositions of the present invention may comprise two or more different and distinct types of porous particles.
  • each type of porous particles provides a specific desorption and/or dissolution rate profile for the biologically active material in a liquid material so as to form a composite desorption and/or dissolution rate profile for the biologically active material.
  • the surface of the inorganic oxide material in particular the surface in the pores, has not been chemically modified.
  • the pores in the inorganic oxide material are open so that the active ingredient in a liquid material can enter the pores and become adsorbed at the surface of the pores, or can leave the pores as to release the active ingredient.
  • the porous inorganic oxide material of the present invention has an oil adsorption of about 100 to about 600 ml/100 g, or of about 100 to about 500 ml/100 g, or of about 100 to about 450 ml/100 g, or of about 100 to about 400 ml/100 g, or of about 150 to about 400 ml/100 g, or of about 200 to about 400 nil/100 g .
  • the oil adsorption values can be measured with standard methodology, in particular by titrating a predetermined quantity of the inorganic oxide materia! with an oil under constant mixing of the oil inorganic oxide material, until the mass shows excess of oil, such as done in ASTM D28 ! ,
  • the inorganic oxide material in the compositions of the present invention is porous.
  • the pores have a mean pore diameter of greater than 5 nm, or from about 5 nm to about 30 nm; or from about 10 nm to about 30 nm.
  • the mean pore diameter is about 20 to about 25 nm.
  • the porous inorganic oxide material has a pore volume of about 0,5 cnrVg or greater, or about 0,6 cm ' Vg or greater, or about 0,7 em 3 /g or greater.
  • the upper limit of the pore volume is about 3.0 cm ' Vg, or about 2.3 cm'Vg.
  • the porous inorganic oxide material has a BET surface area., as measured by nitrogen adsorption, of about 10 ni ' 7g or greater, or about 100 m ' Vg or greater, or of about 200 nr ' - ' g or greater, or of about 300 m ' Vg or greater.
  • the upper limit of the BET surface area is about 1000 m 2 /g, or about B00 Vg, or of about 600 m 2 /g.
  • the BET surface area may range from about 10 to about 1000 m'Vg, or about 100 to about 800 rn 2 ig, or about 150 to about 600 m'Vg, or about 200 to about 500 nrVg, or about 250 to about 400 m'Vg.
  • the porous inorganic oxide material has (i) a mean pore diameter of from about 5 nm to about 30 nm, (si) a pore volume of about 0.7 cm ' Vg or greater, and (iii) a surface area of about 20 to about 500 m 2 /g, or greater, in another embodiment, this porous inorganic oxide material is in the form of particles, which may have a diameter from about 50 ⁇ ? (or about 44 pm) to about 150 ⁇ (or about 149 pm). n a further embodiment this inorganic oxide porous material has an oil adsorption of about 100 to about 500 ml/100 g. Such materials are attractive due to their superior properties in terms of high loading, hulk density, flowability, desorption and dissolution ehareteristies.
  • the measurements of pore volume are generated by
  • the porous inorganic oxide materia? containing a biologically active ingredient in a liquid material may be obtained from porous inorganic oxide material that do not contain a biologically active ingredient in a liquid material, which biologically active ingredient in a liquid material subsequently is added so that it is adsorped by the porous inorganic oxide material .
  • the biologically active ingredient in a liquid material may be added during one or more of the steps of the preparation of the porous inorganic, oxide material.
  • the inorganic oxide can be dispersed in a liquid compound, which is subsequently used as a reaetant or solvent or medium, which forms the biologically active composition of the present invention.
  • the particles of porous inorganic oxide material of the invention may be free- flowing.
  • said particles may have a Carr index of equal or lower than about 25, e.g, a Carr index of about 10 to about 25, In some embodiments the Carr index may be equal or lower than about 15, e.g. a Carr index of about 10 to about 15.
  • the particles of porous inorganic oxide material of the invention may have a Hausner index of about 1 to about 1.4, in particular of about 1 ,2 to about 1.4,
  • the particles of porous inorganic oxide materia! of the invention may have an angle of repose of about 30* to about 45".
  • the Carr and Hausner index values and angles of repose mentioned herein apply to any of the particles of the invention having a w/w ratio of the biologically active ingredient in a liquid material to inorganic oxide inorganic oxide, as specified herein, and in particular where said w/w ratio is between about 0.5 ; 1 to about 2 : 3.
  • the Carr and Hausner index values and angles of repose mentioned herein apply to any of the particles of the invention that are unloaded with biologically active ingredient in a liquid material.
  • a Carr index greater than 25 is considered to be an indication of poor flowability.
  • the present invention concerns a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a iiquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 300 to about 500 rnI/ ⁇ g;.
  • a decrease of the volume or increase in density in particular the particular decreases of volume or increases in density mentioned herein, during a time period of about 3 hrs, in particular of about 2 hrs is observed, after which time period no further decrease occurs and the volume stays substantially constant,
  • the present invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/100 g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0.5 cnvVg or greater; wherein said composition, after mixing the inorganic oxide particles and liquid material, increases in bulk bulk density by at least about 15% after resting, or at least about 20%, or at least about 25%, or at least about 30%, or at least about 35%. or at least about 40% after resting.
  • each of the changes in volume and3 ⁇ 4r density mentioned herein are after loading and resting up to 2 hours depending upon the Iiquid, and even up to 24 or 48 hours.
  • a decrease of the volume or increase in density in particular the particular decreases of volume or increases in density mentioned herein, during a time period of about 3 hrs, in particular of about 2 hrs is observed, after which time period no further decrease occurs and the volume stays substantially constant.
  • the present invention concerns a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 ⁇ about 500 rnl ⁇ g; and (b) pores having an pore volume, as measured by nitrogen porosimetry, of about 0,5 cm 3 /g or greater; wherein, after mixing the inorganic oxide particles and liquid material and then resting for at least 2 hours, at least about 400 mg of the composition may be loaded into a zero size capsule.
  • at least about 410 mg, or at least about 420 mg, or at least about 430 mg of said composition may be loaded into a zero size capsule.
  • the present Invention relates to a composition
  • a composition comprising porous inorganic oxide particles containing a biologically active ingredient in a liquid material, wherein the inorganic oxide particles possess (a) an oil adsorption of about 100 to about 500 ml/i OO g; (b) pores having an pore volume, as measured by nitrogen porosimetry. of about 0.5 cm J g or greater; and (c) a ratio of liquid material to inorganic oxide particles of at least 1 : 1 ; wherein at least 65% of the liquid material is desorbed from the particles upon desorption.
  • At least 70%, or at least 75%, or at least 80%, or at least 85% of the liquid material is desorbed from the particles upon desorption.
  • the liquid material is desorbed under conditions that simulate desorption of liquid material in biological fluids. Such tests are performed by intense mixing in aqueous medium, as set forth in Example 7.
  • liquid form or “liquid material” in relation to biologically active ingredients refers to such ingredients that in themselves are liquids or to biologicaliy active ingredients brought into liquid form by various techniques including, for example, desorption and/or dissolution or conversion into a self-emulsifying drug deliver)' system, Such materials may also include solid biologically active ingredients that are suspended, dispersed or incorporated with liquids.
  • liquid form or “liquid material” refers to biologically active ingredients as such or brought into liquid form that are liquid at room tremperaiure or at physiological temperature, or are liquid at temperatures ranging from about 0 t! C to about 60°C, in partuclar about 10°C to about 5G a C, or about 20°C to about 5 : C, Such materials may be solid at certain conditions (e.g., temperature, concentration, etc.) and liquid under other conditions.
  • the porous inorganic oxide material in accordance with this invention contains a biologically active ingredient in liquid form.
  • the term “contain” means that the porous inorganic oxide material is loaded with a biologically active ingredient, the term “loaded” meaning thai the active ingredient is adsorbed at the surface of the inorganic oxide material including the surface within the pores of the inorganic oxide material. A major part of the active ingredient may be incorporated in the pores of the inorganic oxide material.
  • Such inorganic oxide materials with adsorbed biologically active ingredient are referred as "loaded inorganic oxide materials".
  • the terms “loaded” and “incorporated” in this context arc meant to he equivalent,
  • the w/w ratio of the biologically active ingredient in a liquid material (which comprises the biologically active ingredient itself as well as any added materials in the iiquid) to inorganic oxide is in the range of about 0,5: 1 to about 5: 1 , or of about 0.5: 1 to about 3: 1, or of about 0.5: 1 to about 2: 1, or of about 1 : 1 to about 2: 1 ,
  • the composition comprising porous inorganic oxide material containing a biologically active ingredient in a liquid material may have a bulk density- of at least 450 g/1.
  • said bulk density is in the range of from 450 g 1 to 750 g/1, in particular in the range of from 500 g/1 to 700 g/1, or in the range of from 550 g/i to 650 g/1,
  • the biologically active ingredient in a liquid material is a liquid lipid drug.
  • examples include vitamin A, vitamin E (dl-a-Tocopherol), paracetamol ascorbic acid, sesame oil miglyol, or combinations thereof.
  • the biologically active ingredient in a liquid material takes the form of a solution or dispersion of the biologically active ingredient in a nonvolatile solvent, e.g. having a boiling point of above about 150 C.
  • a nonvolatile solvent e.g. having a boiling point of above about 150 C.
  • examples include glycerin, propylene glycol, liquid polyethylene glycols such as polyethylene glycol 200 and 400, polysorbates such as polysorbate 80, or an oil, Oils that can be used include long-chain triglyceride and medium-chain triglyceride oils with different degrees of saturation; vegetable oils such as olive oil, sunflower oil castor oil, linseed oil and the like; modified or hydro!
  • the active ingredient is preferably in the form of micro- or of nanoparticles.
  • concentration of the biologically active ingredient in the solution or dispersion is in the range of from 1 % to 90 %, or from ... (w w).
  • the biologically active ingredient in a liquid material is a self-emulsifying drag delivery system (SEDDS) comprising an oily/Hpid component, a surfactant cosolvent, and a biologically active ingredient
  • SEDDS self-emulsifying drag delivery system
  • the oily/iipid component is generally a fatty acid ester or a medium/long chain saturated, partially unsaturated or unsaturated hydrocarbon, in liquid, semisolid or solid form at room temperature (e.g., solid lipid nanopartic!es, oily suspensions, submicron lipid emulsions, lipid implants, lipid microtubules, lipid mierobubhies, or lipid microspheres, etc).
  • Examples include mineral oil, vegetable oil, modified or hydrolyzed vegetable oils, silicone oil, lanolin, liposomes, refined animal oil, fatty acids, fatty alcohols, and mono-/ t ⁇ tri- glyeerides, including long-chain triglyceride and medium-chain triglyceride oils with different degrees of saturation and semisynthetic medium-chain triglyceride oils having surfactant properties.
  • oils comprised of one or more medium chain fatty acids esters of propylene glycol such as propylene glycol rnonocaprate, propylene glycol dicaprate, propylene glycol dicaprylate/dicaprate, propylene glycol dipelargonate, and propylene glycol dilaurate, triacetin, fats and oils such as olive oil, sesame oil, soybean oil, corn oil, rape oil, castor oil, coconut oil, and eucalyptus oik .
  • propylene glycol such as propylene glycol rnonocaprate, propylene glycol dicaprate, propylene glycol dicaprylate/dicaprate, propylene glycol dipelargonate, and propylene glycol dilaurate, triacetin, fats and oils such as olive oil, sesame oil, soybean oil, corn oil, rape oil, castor oil, coconut oil, and eucalyptus oik .
  • capryiic/eapric acid triglyceride iglyol ' 812
  • a triglyceride such as tricaprylin and trilaurin
  • po!yglycerm fatty acid esters such as tetraglycerin polyricinoleate, hexaglycerin polyricinoieate, condensed poiyricinoleate, and tetraglycerin mixed fatty acid esters .
  • medium chain fatty acid is meant to refer to fatty acyi chains of between 6 and 14 carbons in length, more preferably between S and 12 carbons in length;
  • long chain fatty acid is meant to refer to fatty acyl chains greater than 14 carbons in length;
  • short chain fatty acid is meant to refer to fatty acyl chains less than 6 carbons in length.
  • the oil components may be used in the SEDDS formulations of the present invention in any effective concentration, including, for example, in a concentration range of 5% to 80% (w/v).
  • Preferred surfactants comprise non-ionic surfactants with a relatively high hydrophiiie-lipophilic balance (HLB) value usually in concentration ranges between 30% and 60% (w/w).
  • HLB hydrophiiie-lipophilic balance
  • the hydrophilic surfactant (HLB (hydrophile-lipophiSe balance) of 9,0 or higher that can be used include polyoxyethylene lauryl ethers (Laureth 2 (BL-2), Laureth 4.2 (BL-4.2), and Laureth 9 (BL-9), polyoxyethylene (20) sorbitan monococonut oil fatty acid ester ("Polysorbate 20"), Polysorbate 40, Polysorbate 80, Labrasol, D-ot -tocopherol polyethylene glycol 1000 succinate (Vitamin E TPGS NF), lauroyl polyoxyethylene glycerin (Gelucire 44/34), polyoxyethylene hydrogenated castor oil 40 , polyoxyethylene hydrogenaied castor oil 60 (HCQ-6G), poiyoxyethylene sorbitan rnonolaurate, poiyoxyethylene sorbitan monopalmitate, and poiyoxyethylene sorbitan monooleate,
  • HLB hydrophilic surfactant
  • Any effective non-aqueous protic coscken or combinations thereof, may be used in the SEDDS for use in the invention.
  • Acceptable non-aqueous proiic solvents include any pharmaceutically acceptable mono-, dk tri-, or poly-hydroxy linear aliphatic and aromatic solvent, or combinations thereof.
  • non-aqueous protic solvents include ethanol, propanol, benzyl alcohol, propylene glycol, liquid polyethylene glycols such as polyethylene glycoi 200 and 400, and glycerol
  • the protic solvents may be used in the formulations of the present invention i any effective concentration, including, for example, in a concentration range of about 5% to about 50% (w/v).
  • a chelating agent and/or a soluble antioxidant may be included in the SEDDS for use in the invention.
  • Chelating agents may be added to enhance the stability of a hydrophobic drag in the SEDDS composition.
  • Suitable optional chelating agents include any pharmaceutically acceptable chelating agent, such as citric acid, nia!eie acid, succinic acid, tartaric acid, EGTA (ethylene giyeol-bis ( (3-aminoethyl ether) tetraaeetic acid, or egtazie acid) and EDTA (ethylene diamine tetraaeetic acid, or edetic acid).
  • Such chelating agents are available in various forms, e. g. 4 as sodium or potassium salts or as the free acids.
  • Such chelating agents may be used in the formulations of the present invention in any effective concentration, including, for example, in a concentration range of between 0,01% and 10% (w/v),
  • an absorption ⁇ promoter such as sodium salicylate, sodium deoxycholate, sodium myristate, or sodium dodecyl sulfate.
  • the SEDDS formulations may contain an auxiliary solvent such as ethanol, propylene glycol, polyethylene glycoi, diethylenetriaminepentaacetic acid, diethanolamine, triethanolamine, ethylenediamine, monoethanoiamine, or N,N-dimethy!acetarnide.
  • an auxiliary solvent such as ethanol, propylene glycol, polyethylene glycoi, diethylenetriaminepentaacetic acid, diethanolamine, triethanolamine, ethylenediamine, monoethanoiamine, or N,N-dimethy!acetarnide.
  • the SEDDS formulation may be prepared by dissolving the drug in a mix of oil, surfactant and cosever.
  • the biologically active material used in the compositions of the present invention may comprise any known biologically active material.
  • biologically active ingredient is meant to cover any pharmaceutical or other active ingredient for administration to humans or animals, in particular to warm-blooded animals.
  • the biologically active material may be an active pharmaceutical ingredient (API), which comprises include naiiirai, semi-synthetic or synthetic moiecules.
  • the biologically active material comprises two or more active pharmaceutical ingredients (APIs) in combination with one another.
  • Other biologically active ingredients include ingredients that have an effect on the general well-being or have an effect on the outer appearance (cosmetic) such as the skin, hair, lips, and eyes.
  • Such ingredients include any agents for use in cleansing, beautifying, promoting attractiveness, or altering the appearance, for example moisturizers, oils, anti-wrinkle agents, fragrances, and the like.
  • ingredients for nutritious applications include food supplements such as, for example, dietary food supplements, vitamins, minerals, fiber, fatty acids, and amino acids. Examples of such ingredients are Vitamin C, omega-3 fatty acids, carotenes, and flavonoids.
  • biologically active in relation to compositions for cosmetic or nutricious applications also includes activity relating to the improvement of the outer part of the body, in particular of the dermis, as well as the general well-being of an individual.
  • the active ingredient has a molecular weight below about
  • the active ingredient for use in the invention may be soluble or insoluble in water or aqueous media, in particular physiological aqueous media.
  • any solvent solubility is defined as the amount of a solvent (g) required to dissolve 1 g of a compound, whereby the following solubility qualifications are defined: 10- 30 g ("soluble”); 30-100 g (“sparingly soluble”); 100-1000 g ("slightly soluble”); 1000-10000 g ("very slightly soluble” or “poorly soluble”) and more than 10000 g (practically insoluble).
  • the active ingredient is soluble or insoluble in water or aqueous media, in particular physiological aqueous media.
  • the pharmaceutically active ingredient belongs to the so-called BCS classes I through IV, Classes I and III are the soluble drugs.
  • the Biopharmaceutical Classification System (BCS) classifies drug substances based on their aqueous solubility and intestinal permeability into four classes: Class i— High Permeability, High Solubility: Class II— High Permeability, Low Solubility: Class Ill-Low Permeability, High Solubility; Class IV-Low Permeability, Low Solubility.
  • the active ingredient has a partition coefficient (expressed as log P) that is in the range from 4 to 9, in the range from 3 and 8. in a further embodiment, the active ingredient has a pK A that allows the molecule to be in neutral (non-ionic form) at about pH 5-8,
  • Exemplary APIs include, aiorvastatin, amiodarone, eandesartan ⁇ cilexetil, : earvedilol, elopidogrel bisulfate, dipyridamole, eprosartan mesylate, epierenone, ezetirnibe, felodipine, furosemide, isradipine, iovastatin, metolazone, nicardipine, nisoldipine, olmesartan medoxornil, propafenone HC1, qinapril, ramipril, simvastatin, telmisartan, trandoiapril, valsartan and other cardio-vascular active drugs; acyclovir, adefovir, dipivoxil, amphotericin, Amprenavir, cefrxirne, ceftazidime, clarithromycin,
  • irinotecan melphalan.
  • mer- captopurine mitotane, paclitaxel, valrubicin, vincristine and other drugs used in oncology
  • azathioprine tacrolimus, cyclosporin, psmecroHrnus, siroHnms and other smmonosupressive drugs
  • clozapine, eniacapone fluphenazine, imipramine, nefazodone, olanzapine, paroxetine, pimozide, sertraline, triazolam, zaleplon, ziprasidone, risperidone, carbamazepine and other drugs for CNS indications
  • danazol dutasteride, medroxyprogesterone, estradiol raloxifene, sildenafil, tadalafi!, testosterone, vardenafii and other drugs used for reproductive health
  • the APIs include ezetimimbe, giucoroniude, tadalafii, fenofibrate, danazol, Itraconazole, carbamazepine, gnseofu!vin, nifedipin.
  • the active ingredients further include sugars, polysaccharides, vitamins, amino acids, peptides, prostaglandins, nucleic acids, nucleotides, nucleosides, as well as derivatives thereof. Also included are peptides, proteins, protein fragments, antibodies, small antibody fragments, and the like. The latter include Fv" fragments, single-chain Fv (scFv) antibodies, antibody Fab fragments, antibody Fab' fragments, antibody fragments of heavy or light chain CDRs, or nanobodies. Also encompassed are small oligonucleic acid or peptide molecules such as aptamers, for example DNA aptamers, RNA aptaraers or peptide aptamers.
  • aptamers for example DNA aptamers, RNA aptaraers or peptide aptamers.
  • the biologically active ingredient in a liquid material when loaded in the inorganic oxide material shows an increased release compared to the active ingredient as such, or to formulations containing the active ingredient and ingredients that do not influence release.
  • Increased release may for example be an increase of 10%, or of 20%, or of 30%, or of 50%, of the weight percentage of active ingredient released under physiological conditions (pH, temperature).
  • the biologically active Ingredient in a liquid material when loaded in the inorganic oxide material shows immediate release from the compositions of the invention
  • immediate release meaning, for example, a release of at least 60% of the drug under physiological conditions (pll, temperature), such as within 60 minutes or less, such as within 30 or less, or within 20 minutes or less, or within 15 minutes or lexs,
  • the step of incorporating the biologically active material into the inorganic oxide materia! typically comprises a variety of loading methods, including the solvent method and the incipient wetness method, which methods have been described in the prior art, or mere mixing without use of any solvent or other mixing aid.
  • the inorganic oxide material is loaded with an aciive ingredient by treatment with a solution of the active ingredient in a liquid material after which the solvent is removed.
  • the active ingredient in a liquid material thereby becomes adsorbed to the surface of the inorganic oxide material, including the surface within the pores of the inorganic oxide material.
  • Appropriate organic solvents for use in this method include dichloromethane, 1 ,4-dioxane, tetrahydrofuran, 2-propanoL diethyl ether, ethyl acetate, acetonitrile, dimethySformamide, N-methyl ⁇ pyrrolidinone, hexane.
  • a solution containing about 50 nig of active ingredient per ml can be used for loading active ingredients in inorganic oxide material.
  • incipient wetness method also referred to as capillary impregnation or dry impregnation the inorganic oxide material is wetted with the active ingredient in a liquid material or in a concentrated solution and is drawn into the pores by capillar ⁇ ' action.
  • porous inorganic oxide materials of the invention are particularly suited for this methodology as they show strong capillary action.
  • no or very little solvent needs to be used thereby avoiding the removal of the solvent after the loading step.
  • This offers an additional advantage over known liquisolid formulations, which require additional ingredients needed to help adsorption, in particular solvents, involving methods of premixing the carrier or the drug in a liquid material (lipid or SEDDS) with a solvent to improve the adsorption.
  • solvents in medicines and other products for human or animal use are critically scrutinized while many solvents are banned. Solvents also have environmental implications as they are considered an important source of pollution.
  • the liquid material may be loaded onto the inorganic oxide material by spraying, or any other known method of liquid adsorption onto porous materials.
  • the biologically active ingredient in a liquid material either with or without solvent may have a viscosity that is selected such that it can be adequately adsorbed by the inorganic oxide material s in particular in terms of speed of adsorption, sufficient loading, and the like, it may for example have a viscosity below about 250 mPa.s, or below about 100 mPa.s, or below about 10 mPa.s, or below about 5 mPa.s, or below about i mPa.s.
  • the lower limit of the viscosity may be about 0.1 mPa.s, or about 0.5 mPa.s.
  • the inorganic oxide materials of the invention are very efficient adsorbants of biologically active ingredient in liquid form. Contrary to known porous materials, where adsorption takes place in a very short time after being brought in contact with the liquid material, the inorganic oxide materials of the invention adsorp biologically active ingredient in liquid form during longer periods of time, in particular during several hours, for example up to about 2, 3, or 4 hours, k is believed that the decrease in volume after resting is related to capillar ⁇ ' farces that continue to draw free liquid between the particles up into the pores of the particles. This attributes to the high loading capacity of the inorganic oxide materials of the invention.
  • compositions of the present invention may in one or more additional steps formulated into a final dosage form, which may vary depending upon the marsner in which it is administered to the patient, Preferred are solid or semisolid dosage forms for oral administration, in particular pills, tablets, and eapsulees. Such dosage forms may be suitable for providing immediate or fast in vivo release of said biologically active species, or may be suitable for controlled release. This may include one or more pharmaceutically acceptable excipients
  • compositions containing a biologically active material and an inorganic oxide material in accordance with this invention may be introduced at any time during the process, including the step designed to load the biologically active material into the pores of the inorganic oxide material, or afterwards in a separate step,
  • compositions may also contain optional excipients.
  • excipients may comprise any of the ingredients customarily employed in the art such as diluents, binding agents, granulating agents, giidants (flow aids), lubricants: disintegrants, sweeteners, flavors, and pigments to make the tablets visually attractive.
  • excipients include hydroxypropylmethyl cellulose, crospovidone, magnesium s earate, lactose, and talc.
  • compositions of the present invention may further comprise one or more pharmaceutically acceptable fillers selected, for example, from hydrocoiioids (such as xantl an gum), binding agents, giidants, lubricants, surfactants and diluents.
  • binding agents such as starch, gelatin, glucose, alginic acid, sodium and calcium alginates, water-soluble acrylic (co) polymers, polyvinylpyrrolidone, polyaminoacids, ethylene-vinyi acetate copolymers and the like; natural and synthetic mineral fillers or giidants such as silica, magnesium silicates such as tale, diatornaceous earth, aluminum silicate such as kaolinite, montmorillonitc or mica, magnesium aluminum silicate such as attapulgite and vermieulite, carbon such as charcoal, sulphur and highly dispersed silicic acid polymers; water-soluble diluents such as lactose, sorbitol and the like.
  • binding agents such as starch, gelatin, glucose, alginic acid, sodium and calcium alginates, water-soluble acrylic (co) polymers, polyvinylpyrrolidone, polyaminoacids, ethylene-vinyi acetate copolymers and
  • compositions of the present invention may also be formulated into forms suitable for topical application such as an ointment, a cream, a gel, a liniment or balm, etc...
  • the present invention is further directed to methods of using any of the herein disclosed compositions.
  • the compositions, in particular the pharmaceutical compositions, of the present invention may be used as medicaments, in particular may be used as medicaments via the oral route.
  • the present invention a method of administering a composition to a patient so as to deliver at least one biologically active material to the patient wherein the composition comprises at least one pharmaceutical dosage formulating ingredient of a porous inorganic oxide material containing a biologically active ingredient in liquid form, wherein the inorganic oxide material has the oil adsorption, the intraparticie pore volume, and BET surface area, as specified herein.
  • the compositions in this method are preferably administered by various means, including by oral, buccal, sublingual, periodontal, vaginal intrauterine, rectal, pulmonary, nasal, inhalation, intraocular, ophthalmic, auricular, and topical means,
  • One of the reasons for improved release of the biologically active material from the compositions of the present invention is due to the improved desorption of the liquid material from the inorganic oxide material.
  • the presence of pores within the inorganic oxide materials i.e., intraparticie porosity
  • the pore size distribution of the present inorganic oxide material is narrow (i.e., a small relative span), which allows for a number of pores to readily adsorb and desorb the liquid material.
  • compositions in accordance with the invention provide attractive drug delivery properties. They provide desirable desorption and/or dissolution rate profiles for a variety of biologically active materials (e.g., APIs), in some embodiments, the biologically active material exhibits a percent release desorption and/or dissolution rate of about 20 or greater within about 15 minutes of an initial time of contact with a dissolution medium. In some embodiments, the biologically active material exhibits a percent release dissolution rate of about 25 or greater (or about 30 or greater; or about 35 or greater) within about 15 minutes of an initial time of contact with a dissolution medium.
  • biologically active materials e.g., APIs
  • the biologically active material exhibits a percent release desorption and/or dissolution rate of about 20 or greater within about 15 minutes of an initial time of contact with a dissolution medium. In some embodiments, the biologically active material exhibits a percent release dissolution rate of about 25 or greater (or about 30 or greater; or about 35 or greater) within about 15 minutes of an initial time of contact with a dissolution medium
  • the biologically active material exhibits a percent release dissolution rate of about 20 or greater about 30 minutes after an initial time of contact with a dissolution medium, in some embodiments, the biologically active material exhibits a percent release dissolution rate of about 30 or greater about 30 minutes after an initial time of contact with a dissolution medium.
  • the biologically active material exhibits a percent release dissolution rate of about 10 or greater about 60 minutes after an initial time of contact with a dissolution medium. In some embodiments, the biologically active material exhibits a percent release dissolution rate of about I S or greater (or about 20 or greater) about 60 minutes after an initial time of contact with a dissolution medium,
  • compositions of the present invention in many instances show immediate release of the active ingredient but may be turned into controlled release compositions for example by coating the compositions with a suitable polymer.
  • a suitable polymer When mixing compositions with sleeted polymer coatings mixed release patterns can be obtained such as a combination of immediate and sustained release.
  • the inorganic oxide material may comprise two or more different and distinct types of porous particles with each distinct type of porous particles providing a specific desorption and/or dissolution rate profile for a single biologically active material (or two or more different biologically active materials) so as to form a composite desorption and/or dissolution rate profile.
  • a further aspect of this invention concerns particles of a inorganic oxide material, wherein the inorganic oxide material wherein the inorganic oxide material has the oil adsorption, the intraparticle pore volume, and BET surface area, as specified herein.
  • compositions of the present invention not only show high loadabiiity of drugs in liquid form, they moreover show higher bulk density compared to existing Siquisolid systems. Further favorable properties include the excellent adsorptive capacity of the inorganic oxide material of the invention and the increased stability of the active ingredient. These advantages are in particular offered by the compositions of the invention in the form of particles. [0138]
  • the compositions of the present invention can also be used in dermatology and cosmetic applications because of their good skin-comparability and lack of unpleasant skin feel
  • the DI G, D50 and D90 values indicate the 10 th , 50 th and 90 th percentiles of the weight of the particle diameter distribution. These values were obtained from a Malvern f M rV!astersizer 2000 instrument available from Malvern Instruments Ltd. PV: pore volume; SA: surface area; APD: average pore diameter; BET SA: surface area; and Relative Span are determined using BJH nitrogen adsorption at a pressure of 0.995 using an ASAP 2420HV accelerated surface area and porosimeiry system available from Micromeretics Instrument Corporation.
  • Example 3 The Cremophor and Labrafii loaded materials of Example 3 were loaded into zero size capsules manually using a casule filling tray, such as the Cap- -Quick available from Empty Caps Company. As can be seen from FIG, 2, there ts a significant difference between the filling amounts before and after resting of the material The amounts of material loaded into the capsules are set forth in Tables 8 and 9.
  • the ascorbic acid loaded carrier (S i ) was subjected to dissolution studies as per USP 30 for Ascorbic acid Tablets for 45 min.
  • Dissolution test conditions comprised of use of USP dissolution apparatus 2 (Paddle) operated at speed of 50 PM for 45 minutes.
  • Dissolution medium was 900 ml of water at 37 ⁇ Q,5 C.
  • 100 rng of the weighed amount of drug loaded carrier was used to dissolution studies.
  • Aliquots (5 ml) were withdrawn at 10, 20, 30 minute time intervals, filtered and diluted with water. Absorbance of aliquots was determined spectromeiricaliy at X ax 266nm.
  • the release profile is illustrated in FIG, 4.
  • the drug release from the both carriers S i and S2 met the USP criteria (NLT 75% in 45 minutes).
  • Oil release or desorption The carrier material used in the following trials was the material designated S I prepared in Example 1 , 2 grams of oil loaded carrier ( 1 : 1 , w/w) prepared pursuant to method of Example 2 was mixed with 6 ml water in a beaker, vorlexed during 1 hour, and centrifuged at 5000 RPM for 10 min in a Heraeus Muitifuge 1 S ⁇ R centrifuge available from Thermo Electron Corporation. The supernatant, i.e. oil + water, was transferred into a petry dish and dried in hot air oven up to constant weight.
  • the results which are based upon the w/w % release obtained in the trials., are as follows. For sesame oil, 81 % of the oil is released from the carrier materia! or inorganic oxide, and tor Miglyol 812, 81.3% of the oil is released from the carrier material or inorganic oxide.
  • Solid SEDDS System Loading and Release (or Desorption) The carrier materials used in the following trials was the materials designated as SI and S2 in Example 1.
  • a liquid SEDDS system was made up containing 0,6g of Giyburide as the API component, I Sg of Capryol®90 as the oil/vehicle component. 54.4g of Trascutoi® HP as a co-surfactan and 30g of Tween® 20 as a surfactant.
  • This liquid SEDDS system was loaded onto SI and S2 by accurately weighing the required quantities of carrier and liquid SEDDS in the ratio of 1 : 1.
  • Carrier and Liquid SEDDS were pre-heated at 60 °C for S 5 min, prior to mixing. Liquid SEDDS was added slowly to the carrier under stirring with metallic spatula. The prepared mixture was kept aside for around 24 hr to get free flowing powder.
  • Dissolution test conditions comprised of use of USP dissolution apparatus 2 (paddle) operated at a speed of 75RPM.
  • Dissolution medium was 500ml of pH 9.5 Borate Buffer (0.05M) at 37 ⁇ 1°C.
  • a weighed amount of ioaded carrier or APi(non- micronized Gyburide) with an weight equivalent basis of 5rng Gyb ride were used for the dissolution studies.
  • Cremophor® EL was the liquid lipid that was loaded on to the silica.
  • Silica 52 was loaded pursuant to method of Example ⁇ and was used as the oil loaded carrier component in tablet formulations. To deliver oil in tablet dosage form, tablets were made with two different processes: direct compression (DC) and wet granulation (WG).
  • DC direct compression
  • WG wet granulation
  • Direct compression tablets were obtained by accurately weighed the quantities of excipients for blend preparation.
  • Diluent (MCC ) and Oil loaded carriers were sieved through #40 mesh and mixed well for approximately 5 min. Binders and disiniegrant were sieved through #40 mesh and added to the blend, then mixed well for approximately 5 min.
  • GHdant was passed through #40 mesh and added to the blend and mixed well for 5 min.
  • Lubricant was sieved through # 60 mesh and added to the blend and mixed well for approximately 2min. This final blend was used for the compression of the tablets.
  • Ghdant was passed through #40 mesh and added to the blend and mixed well for 5 min.
  • Lubricant (what material) was sieved through # 60 mesh and added to dried granules and mixed well for approximately 2mirt. This final blend was used for the compression of the tablets.
  • Tablet hardness for all formulations was measured with EH 01 tablet hardness tester (Eleetrolab, India). The hardness for tested formulations are listed in Table Y below.
  • Example 9 Additional excipients were added pursuant to the method in Example 9 to obtain a direct compression blend with the following composition : 70% Tocopherol loaded silica S2(l : 1 loaded), 12.5%MCCPH102, 34% PVP30, 2% AcDiSoi, 1 % SYLOSD® 244FP, 0.5% Magnesium Stearate. Tablets were prepared using a Parle Elizabeth tools Pvt Ltd, Eliza press 200 mu!ti tooling single rotary tablet press operated at a Speed of 5 rprn, compression force of 20kN, ejection force ⁇ 70N, and with a 12mm round bioconeave, D-tao!ing punch.
  • Tablet hardness for all formulations was measured with EH 01 tablet hardness tester (Eieetrolab. India) at tablet weight of 5GiK-5rng. Tablet hardenss results for these tablets was 40N. Friability was 0% and disintegration time was ⁇ l min.
  • Oil Release (Tocopherol) from Tablets The carrier materials used in the following trials was the material designated S2 in Example L Tocopherol was the liquid lipid that was loaded on to the silica. Silica S2 was loaded with liquid lipid pursuant to method of Example 8 and was used as the oil loaded carrier component in tablet. Tablets were prepared pursuant to the method in example 10. Tochopherol concentration in the tablet was 1 OOmg. [0168] Tablets prepared were .subjected to dissolution studies pursuant to the method described in Example 8. An aliquot of 2 mL was withdrawn at predetermined time interval and filtered through 0.22 ⁇ membrane filter. The dissolution samples were analyzed by using HPLC (Waters UPLC Wavelength: 294 rtm Column: Rocket Format, 53x7mm, 3 ⁇ ) Mobile Phase: 85% ACN: 10% MeOH: 5% H20.)
  • any number R falling within the range is specifically disclosed, in particular, the following numbers R within the range are specifically disclosed: R ;; R L + k(Ru -RL) > where k is a variable ranging from ! % to 100% with a 1% increment e.g., k is 1 %, 2%, 3%, 4%, 5%. ... 50%, 51 , 52%. , , , 95%, 96%, 97%, 98%, 99%, or 100%.
  • any numerical range represented by any two values of , as calculated above is also specifically disclosed.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Health & Medical Sciences (AREA)
  • Inorganic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Silicon Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Solid-Sorbent Or Filter-Aiding Compositions (AREA)

Abstract

L'invention concerne des compositions contenant un principe biologiquement actif et une matière d'oxyde inorganique. Des procédés de fabrication et d'utilisation des compositions contenant un principe biologiquement actif et une matière d'oxyde inorganique sont également décrits. La présente invention concerne des compositions comportant une matière poreuse d'oxyde inorganique contenant un principe biologiquement actif sous forme liquide, des procédés de fabrication de telles compositions et leurs procédés d'utilisation.
PCT/US2014/013848 2013-02-01 2014-01-30 Gel de silice poreux comme support pour des technologies liquides WO2014120922A1 (fr)

Priority Applications (8)

Application Number Priority Date Filing Date Title
ES14746507T ES2908958T3 (es) 2013-02-01 2014-01-30 Gel de sílice porosa como portador para tecnologías líquidas
CN201480019478.7A CN105102117A (zh) 2013-02-01 2014-01-30 作为用于液体技术的载体的多孔硅胶
PL14746507T PL2950923T3 (pl) 2013-02-01 2014-01-30 Porowaty żel krzemionkowy jako nośnik płynnych technologii
KR1020157023665A KR102229036B1 (ko) 2013-02-01 2014-01-30 액체 기술을 위한 담체로서의 다공성 실리카 겔
US14/764,363 US10660856B2 (en) 2013-02-01 2014-01-30 Porous silica gel as a carrier for liquid technologies
EP14746507.4A EP2950923B1 (fr) 2013-02-01 2014-01-30 Gel de silice poreux comme support pour des technologies liquides
JP2015556128A JP6513031B2 (ja) 2013-02-01 2014-01-30 液体テクノロジーのための担体としての多孔質シリカゲル
HK16105942.0A HK1217927A1 (zh) 2013-02-01 2016-05-25 作爲用於液體技術的載體的多孔硅膠

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361759723P 2013-02-01 2013-02-01
US61/759,723 2013-02-01

Publications (1)

Publication Number Publication Date
WO2014120922A1 true WO2014120922A1 (fr) 2014-08-07

Family

ID=51262940

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/013848 WO2014120922A1 (fr) 2013-02-01 2014-01-30 Gel de silice poreux comme support pour des technologies liquides

Country Status (9)

Country Link
US (1) US10660856B2 (fr)
EP (1) EP2950923B1 (fr)
JP (2) JP6513031B2 (fr)
KR (1) KR102229036B1 (fr)
CN (2) CN111389345A (fr)
ES (1) ES2908958T3 (fr)
HK (1) HK1217927A1 (fr)
PL (1) PL2950923T3 (fr)
WO (1) WO2014120922A1 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3445337B1 (fr) * 2016-04-22 2021-03-24 University Of Mississippi Granulation à sec à double vis pour la production de formulations solides
BR112020004197A2 (pt) * 2017-09-01 2020-09-08 Jw Pharmaceutical Corporation preparação sólida compreendendo dutasterida e método para preparar a mesma
CA3114751A1 (fr) 2018-10-05 2020-04-09 Fuji Chemical Industries Co., Ltd. Composition de particules de silice poreuse
CN111751480B (zh) * 2020-07-16 2021-06-15 北京理工大学 一种维生素e琥珀酸聚乙二醇酯的应用
CN116159031B (zh) * 2023-02-22 2023-08-22 上海国创医药股份有限公司 一种马来酸氟伏沙明片剂及其制备方法

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4157920A (en) 1975-03-12 1979-06-12 J. M. Huber Corporation Novel precipitated siliceous products and methods for their use and production
US4226743A (en) 1979-03-12 1980-10-07 W. R. Grace & Co. Silica-alumina hydrogel catalyst
US20030152771A1 (en) * 1999-04-22 2003-08-14 Preston Barry W. Very high structure, highly absorptive hybrid silica and methods for making same
WO2004073689A1 (fr) 2003-02-19 2004-09-02 Lifecycle Pharma A/S Utilisation de silice ou d'un derive de silice en tant que materiau de sorption
US20050112232A1 (en) * 2002-04-30 2005-05-26 Jean-Marie Dollat Animal feed supplement
US20090311159A1 (en) * 2006-08-22 2009-12-17 Evonik Degussa Gmbh Fumed silica for use as auxiliary in pharmaceutical and cosmetic compositions
WO2011144346A1 (fr) * 2010-05-21 2011-11-24 Grace Gmbh & Co. Kg Particules poreuses d'oxyde inorganique et leurs procédés de fabrication et d'utilisation
US20120269792A1 (en) * 2001-11-14 2012-10-25 Jarrow Formulas, Inc. Eutectic-based self-nanoemulsified drug delivery system

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4629588A (en) * 1984-12-07 1986-12-16 W. R. Grace & Co. Method for refining glyceride oils using amorphous silica
US5256386A (en) * 1987-06-29 1993-10-26 Eka Nobel Ab Method for preparation of silica particles
DE533865T1 (de) * 1991-03-16 1993-11-04 Torf Establishment, Vaduz Torf-derivierte bioaktive produkte und diese enthaltende pharmazeutische und kosmetische zusammensetzungen.
DE4429644A1 (de) * 1994-08-20 1996-02-22 Sued Chemie Ag Iodadsorptionsmittel
US5800834A (en) 1996-06-10 1998-09-01 Spireas; Spiridon Liquisolid systems and methods of preparing same
US6096337A (en) 1996-06-10 2000-08-01 Spireas; Spiridon Liquisolid systems and methods of preparing same
JPH10130013A (ja) * 1996-09-09 1998-05-19 Toyota Central Res & Dev Lab Inc クラスター包接材料
FR2767071B1 (fr) * 1997-08-06 1999-09-10 Rhodia Chimie Sa Composition comprenant un liquide absorbe sur un support a base de silice precipitee
JPH11157827A (ja) * 1997-11-21 1999-06-15 Shionogi & Co Ltd 新規二酸化ケイ素
SE9704400D0 (sv) 1997-11-28 1997-11-28 Astra Ab Porous inorganic particles as carriers for drug substances
US6342249B1 (en) 1998-12-23 2002-01-29 Alza Corporation Controlled release liquid active agent formulation dosage forms
US20030236236A1 (en) * 1999-06-30 2003-12-25 Feng-Jing Chen Pharmaceutical compositions and dosage forms for administration of hydrophobic drugs
US6790814B1 (en) 1999-12-03 2004-09-14 Procter & Gamble Company Delivery system having encapsulated porous carrier loaded with additives, particularly detergent additives such as perfumes
GB2365769A (en) 2000-08-18 2002-02-27 Secr Defence Skin preparations containing silicon
DE10153078A1 (de) * 2001-10-30 2003-05-22 Degussa Verwendung von Granulaten auf Basis von pyrogen hergestelltem Siliciumdioxid in pharmazeutischen Zusammensetzungen
DE10244397A1 (de) 2002-09-24 2004-04-01 Basf Ag Cholinascorbat-Formulierungen
FR2850576B1 (fr) 2003-02-05 2007-03-23 Ethypharm Sa Composition comprenant un melange de principes actifs, et procede de preparation
FR2851918B1 (fr) 2003-03-06 2006-06-16 Poudre impregnee ameliorant la biodisponibilite et/ou la solubilite et procede de fabrication
ATE371434T1 (de) 2003-03-27 2007-09-15 Dow Corning Zusammensetzungen mit verzögerter freisetzung
GB0312703D0 (en) 2003-06-03 2003-07-09 Oxonica Ltd Agricultural compositions
JP2005053728A (ja) * 2003-08-01 2005-03-03 Dsl Japan Co Ltd 高吸油性および高い構造性を有する非晶質シリカ粒子
JP2005053744A (ja) * 2003-08-05 2005-03-03 Dsl Japan Co Ltd 高吸油性非晶質シリカ粒子
JP5390739B2 (ja) * 2003-09-11 2014-01-15 太陽化学株式会社 物質担持多孔質シリカ
EP1677828B1 (fr) 2003-10-21 2012-12-26 PSIMedica Limited Materiau composite comprenant un semi-conducteur poreux impregne avec au moins une substance organique
GB0406819D0 (en) 2004-03-26 2004-04-28 Dow Corning Controlled release compositions
JP2005307120A (ja) * 2004-04-26 2005-11-04 Mitsubishi Chemicals Corp 徐放剤及びそれを用いた徐放材
GB0420016D0 (en) * 2004-09-09 2004-10-13 Leuven K U Res & Dev Controlled release oral delivery system
CN100352430C (zh) * 2005-03-09 2007-12-05 山东大学 姜黄素自微乳化制剂及其制备方法
WO2006119779A2 (fr) 2005-05-10 2006-11-16 Lifecycle Pharma A/S Composition pharmaceutique comprenant un antagoniste de l'aldosterone
FR2894822B1 (fr) 2005-12-20 2011-11-18 Pf Medicament Composition pharmaceutique contenant des acides gras omega-3
US8920821B2 (en) 2006-04-14 2014-12-30 Perrigo Israel Pharmaceuticals Ltd. Pharmaceutical compositions comprising silica microspheres
CA2649870C (fr) 2006-05-04 2014-07-29 University Of South Australia Liberation de medicaments a partir de gelules recouvertes de nanoparticules
US7691400B2 (en) 2006-05-05 2010-04-06 Medtronic Vascular, Inc. Medical device having coating with zeolite drug reservoirs
KR20080026754A (ko) * 2006-09-21 2008-03-26 주식회사 삼양사 생물학적 활성성분을 함유하는 방출 제어형 입자, 및 이의제조방법
US20100136124A1 (en) 2007-04-20 2010-06-03 University Of South Australia Nanoparticle-coated capsule formulation for dermal drug delivery
EP2216015A4 (fr) 2007-11-02 2012-12-19 So Pharmaceutical Corp Produit complexe de substance médiocrement soluble-agent tensio-actif, et son procédé de fabrication
EP2072042A1 (fr) * 2007-12-21 2009-06-24 Lek Pharmaceuticals D.D. Ingrédient pharmaceutique actif sur support solide amorphe et avec solubilité améliorée
KR100950548B1 (ko) * 2008-01-10 2010-03-30 연세대학교 산학협력단 다공성 중공 실리카 나노입자, 그의 제조 방법, 상기를포함하는 약물 전달체 및 약제학적 조성물
DE102008000290A1 (de) 2008-02-13 2009-08-20 Evonik Degussa Gmbh Lagerstabile Produktsyteme für Prämixformulierungen
EP2251038B1 (fr) 2008-03-11 2017-05-10 ASKA Pharmaceutical Co., Ltd. Dispersion solide, compositions pharmaceutiques comprenant celle-ci, et procédés de production associés
US8188022B2 (en) 2008-04-11 2012-05-29 Amcol International Corporation Multilayer fragrance encapsulation comprising kappa carrageenan
US8778400B2 (en) 2008-04-21 2014-07-15 University Of South Australia Nanoparticle-stabilized capsule formulation for treatment of inflammation
AU2009282051A1 (en) 2008-08-12 2010-02-18 Castle Beach, L.L.C. Active delivery system formulations
GB0821928D0 (en) 2008-12-01 2009-01-07 Isis Innovation Antibiotic nanomaterial
DE102010003204A1 (de) * 2010-03-24 2011-12-15 Evonik Degussa Gmbh Grobteilige Trägerkieselsäuren
NO2613784T3 (fr) 2010-09-07 2018-05-12
US9980481B2 (en) 2012-04-02 2018-05-29 Basf Se Granules obtainable by milling pesticide and silica, addition of adjuvant, and fluidized bed granulation

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4157920A (en) 1975-03-12 1979-06-12 J. M. Huber Corporation Novel precipitated siliceous products and methods for their use and production
US4226743A (en) 1979-03-12 1980-10-07 W. R. Grace & Co. Silica-alumina hydrogel catalyst
US20030152771A1 (en) * 1999-04-22 2003-08-14 Preston Barry W. Very high structure, highly absorptive hybrid silica and methods for making same
US20120269792A1 (en) * 2001-11-14 2012-10-25 Jarrow Formulas, Inc. Eutectic-based self-nanoemulsified drug delivery system
US20050112232A1 (en) * 2002-04-30 2005-05-26 Jean-Marie Dollat Animal feed supplement
WO2004073689A1 (fr) 2003-02-19 2004-09-02 Lifecycle Pharma A/S Utilisation de silice ou d'un derive de silice en tant que materiau de sorption
US20090311159A1 (en) * 2006-08-22 2009-12-17 Evonik Degussa Gmbh Fumed silica for use as auxiliary in pharmaceutical and cosmetic compositions
WO2011144346A1 (fr) * 2010-05-21 2011-11-24 Grace Gmbh & Co. Kg Particules poreuses d'oxyde inorganique et leurs procédés de fabrication et d'utilisation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2950923A4

Also Published As

Publication number Publication date
US20150366805A1 (en) 2015-12-24
ES2908958T3 (es) 2022-05-04
KR20150115860A (ko) 2015-10-14
JP2019048819A (ja) 2019-03-28
EP2950923A4 (fr) 2016-10-19
HK1217927A1 (zh) 2017-01-27
JP6513031B2 (ja) 2019-05-15
CN105102117A (zh) 2015-11-25
KR102229036B1 (ko) 2021-03-17
JP6861679B2 (ja) 2021-04-21
EP2950923B1 (fr) 2022-03-09
JP2016508502A (ja) 2016-03-22
US10660856B2 (en) 2020-05-26
EP2950923A1 (fr) 2015-12-09
CN111389345A (zh) 2020-07-10
PL2950923T3 (pl) 2022-04-04

Similar Documents

Publication Publication Date Title
JP6861679B2 (ja) 液体テクノロジーのための担体としての多孔質シリカゲル
Tan et al. Silica-lipid hybrid (SLH) microcapsules: a novel oral delivery system for poorly soluble drugs
EP2919903B1 (fr) Compositions contenant un matériau biologiquement actif et un oxyde inorganique non ordonné
JP2015521175A (ja) カンナビノイド含有粒状体、その製造方法、及びこのような粒状体を含む経口投与単位
Choudhari et al. Comparative evaluation of porous silica based carriers for lipids and liquid drug formulations
Midha et al. Prospectives of solid self-microemulsifying systems in novel drug delivery
Shazly et al. Dissolution improvement of solid self-emulsifying drug delivery systems of fenofibrate using an inorganic high surface adsorption material
CN108079308A (zh) 辅酶q10与二十八烷醇复配的纳米结构脂质载体及制备方法
CN107921017A (zh) 使用卡多曲组合物进行治疗的方法
US11484501B2 (en) Mechanochemical activated dry amorphisation by milling equilibrium between ap mesoporous silica
Singh et al. Polymeric delivery systems for poorly soluble drugs
US20220354791A1 (en) Mesoporous polymeric particulate material
Hasan et al. Effect of Self-Microemulsifying Lipid Formulations on the Dissolution and Compaction Profiles of Tablets Containing Theophylline; A BCS Class I Compound
LETCHMANAN ENHANCING BIO-PHARMACEUTICAL PROPERTIES OF POORLY SOLUBLE ANTI-MALARIAL DRUGS BY FORMULATING AMORPHOUS FORMS WITH BIOCOMPATIBLE EXCIPIENTS

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201480019478.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14746507

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2014746507

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2015556128

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20157023665

Country of ref document: KR

Kind code of ref document: A